Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Anne Vaslin.
Cancer Research | 2015
Valerie Nicolas-Metral; Anne Vaslin; Jeffrey G. Supko; Kiyohiko Nakai; Nobuya Ishii; Annick Menetrey; Marie-Claude Roubaudi-Fraschini; Judith Marfurt; Sebastien Chabaud; Andreas Layer; Daniela Purcea; Jerome Douchain; Claudio Zanna
Background: Deregulated fibroblast growth factor receptor (FGFR) signaling is associated with tumorigenesis. The oral selective FGFR 1, 2, 3 inhibitor Debio 1347, a Biopharmaceutical Classification System Class II drug, is currently investigated in a phase I trial in selected patients harboring FGFR genetic alterations (NCT01948297). A formulation switch from capsules to tablets was investigated in the course of the dose escalation part of the trial. Methods: Comparative in vitro dissolution tests and PK data after single dose (20 mg) in Cynomolgus monkeys were generated with capsules and tablets to evaluate the suitability of the new tablet formulation prior to human use. The first-in-human, phase I dose-escalation multiple tumor type “basket” study enrolled patients with advanced solid malignancies harboring defined activating alterations of FGFR 1, 2, or 3. Patients received Debio 1347 orally once daily and were assessed for dose-limiting toxicities (DLT) during the first 4 weeks. With a starting dose level of 10 mg the study followed a 3+3 algorithm with dose-escalation on a modified Fibonacci sequence. Capsules of 10- and 20-mg strength were administered in patients up to the 3rd dose level. At the 4th dose-level, the pharmacokinetic (PK) profile and intra-patient relative oral bioavailability of the new tablet formulation versus the original capsule formulation were determined in a two-period, 7-day washout, cross-over design after single dose in standardized food intake conditions. Debio 1347 plasma levels were measured using a validated LC-MS/MS assay. The pharmacodynamic (PD) profile of Debio 1347 was also assessed by measuring plasma levels of several biomarkers using standardized assays. A comparison of the PK and PD of the two dosage forms and their tolerability was performed prior to selection of the dosage form for pursuing the dose escalation. Results: Adequate in vitro dissolution profiles were observed for both capsules and tablets. In addition, oral bioavailability of 20-mg tablets was comparable to that of 10- and 20-mg capsules in monkeys. In a majority of solid tumor patients administered with 40 mg of Debio 1347, intra-patient relative oral bioavailability of the tablet versus capsule was > 80%. In addition, PD and safety profiles after 4-week once daily dosing with tablets were comparable to that of capsules. Conclusion: A formulation switch from capsules (only 10 and 20 mg capsules available) to tablets (20, 30, 50, and 100 mg tablets) of the FGFR inhibitor Debio 1347 was successfully implemented in the course of the first-in-human trial to facilitate dose escalation and improve treatment compliance at high doses by reducing the number of units to be swallowed by solid tumor patients. Citation Format: Valerie Nicolas-Metral, Anne Vaslin, Jeffrey G. Supko, Kiyohiko Nakai, Nobuya Ishii, Annick Menetrey, Marie-Claude Roubaudi-Fraschini, Judith Marfurt, Sebastien Chabaud, Andreas Layer, Daniela Purcea, Jerome Douchain, Claudio Zanna. Formulation switch and pharmacokinetics/pharmacodynamics of Debio 1347 (CH5183284), a novel FGFR inhibitor, in a first-in-human dose escalation trial in solid tumors patients. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr CT228. doi:10.1158/1538-7445.AM2015-CT228
Cancer Research | 2017
Franck Brichory; Anna Pokorska-Bocci; Paolo Nuciforo; Stefania Rigotti; Nathalie Lembrez; Grégoire Vuagniaux; Corinne Moulon; Anne Vaslin
Dysregulation of the fibroblast growth factor receptor (FGFR) signaling pathway due to receptor overexpression, gene amplification, point mutations or fusions/chromosomal translocations is associated with cancer development and progression. Debio 1347 (CH5183284) is an oral selective FGFR inhibitor (FGFR1, 2 and 3) currently in clinical development. The aim of this study was to investigate Debio 1347 activity in patient derived xenograft (PDX) mouse models harboring diverse FGFR alterations in multiple indications. The trial was conducted in 66 PDX models of diverse histotypes selected according to their FGFR1, 2 and 3 alteration status. Debio 1347 was administered orally once daily at 40 up to 80 mg/kg for 10 to 22 consecutive days (N=3/group). Tumor volume was compared to the vehicle control group and measured by caliper twice weekly. Treatment response was determined by relative treatment-to-control ratios (ΔT/ΔC) of which a responding model was defined as ΔT/ΔC Debio 1347 induced tumor regression in 33% of PDX models that exhibited a gene copy number gain and/or the presence of a FGFR fusion gene and/or an FGFR mutation. In addition, Debio 1347 treatment led to tumor regression in 29% of models that did not harbor any FGFR genetic alteration. In contrast, all models that responded to Debio 1347 were shown to display a high expression level (mRNA) of at least one FGFR gene. These findings suggest that high expression of at least one FGFR might be a better predictor of sensitivity to Debio 1347 than genetic alteration. Furthermore, response to Debio 1347 was associated with a decrease in DUSP6 mRNA levels, suggesting that it could be a reliable pharmacodynamic biomarker in clinical trials. These results provide new mechanistic insights into the predictive sensitivity to Debio 1347 and will help refine patient selection for FGFR-targeted therapy. Citation Format: Franck Brichory, Anna Pokorska-Bocci, Paolo Nuciforo, Stefania Rigotti, Nathalie Lembrez, Gregoire Vuagniaux, Corinne Moulon, Anne Vaslin. The activity of the FGFR selective inhibitor Debio 1347 is correlated with high mRNA expression [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2088. doi:10.1158/1538-7445.AM2017-2088
Cancer Research | 2016
Brichory Franck; Anna Pokorska-Bocci; Paolo Nuciforo; Stefania Rigotti; Nathalie Lembrez; Grégoire Vuagniaux; Corinne Moulon; Anne Vaslin
Dysregulation of the fibroblast growth factor receptor (FGFR) signaling pathway due to receptor overexpression, gene amplification, point mutations or fusions/chromosomal translocations is associated with cancer development and progression. In gastric cancer, alterations of FGFR2 are observed in a subset of patients, overexpression being more frequently observed as compared to FGFR2 amplifications. In addition, FGFR2 amplification and ERBB2, EGFR, MET and KRAS amplifications are mutually exclusive suggesting that FGFR targeted therapy would be most effective in the FGFR2-amplified subgroup of patients. Debio 1347 (CH5183284) is an oral selective FGFR inhibitor currently in clinical development. The aim of the study was to investigate the preclinical activity of Debio 1347 in patient derived xenograft (PDX) mouse models harboring diverse FGFR alterations in multiple indications, including gastric cancer. The mouse trial was conducted in 39 different PDX models selected according to FGFR1, 2 and 3 alterations status. Debio 1347 was administered orally once daily and tumor volume was monitored for 14 consecutive days. PDX tumors were extensively characterized using FISH, RNAseq, nCounter Gene Expression and immunohistochemistry (IHC), and FGFR molecular alterations were correlated with Debio 1347 efficacy. The level of tumor heterogeneity for FGFR2 amplification and expression and its impact on drug response were also assessed. In gastric cancer, Debio 1347 induced tumor regression in models harboring the commonly found FGFR2 amplification. Interestingly, Debio 1347 was also highly effective in models with high FGFR2 expression but without FGFR2 amplification, indicating that not only gene amplification drives the activity of Debio 1347 in this indication, but also the expression level of FGFR2. Altogether, these data demonstrate that Debio 1347 could be highly effective in gastric cancer patients harboring FGFR2 amplification and/or high FGFR2 expression. Debio 1347 is currently investigated in a Phase I trial in selected patients harboring FGFR alterations (NCT01948297). Citation Format: Brichory Franck, Anna Pokorska-Bocci, Paolo Nuciforo, Stefania Rigotti, Nathalie Lembrez, Gregoire Vuagniaux, Corinne Moulon, Anne Vaslin. FGFR-selective inhibitor Debio 1347 induces tumor regressions in FGFR2-altered gastric cancer PDX models. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4784.
Cancer Research | 2015
Anne Vaslin; Stefania Rigotti; Nathalie Lembrez; Grégoire Vuagniaux; Corinne Moulon; Hiroaki Tanaka
Dysregulation of the fibroblast growth factor receptor (FGFR) signaling pathway due to receptor overexpression, gene amplification, point mutations or fusions/chromosomal translocations is associated with cancer development and progression. FGFR gene fusions have recently been discovered in several indications including bladder cancer, glioblastoma, lung squamous cell cancer and cholangiocarcinoma. Predominant gene fusions have been identified in some indications such as FGFR3-TACC3 (with different genomic variations in the two genes) for bladder cancer whereas for other indications the genomic-altered landscape is a lot more heterogeneous, as seen in cholangiocarcinoma where at least ten FGFR2 fusions each with a different fusion partner have been identified. We herein report for the first time the discovery of two novel FGFR fusions in intra hepatic cholangiocarcinoma patient samples. These new fusions contain an intact FGFR2 tyrosine kinase domain and fused partner harbor oligomerization domains that suggest similar mechanism of constitutive kinase activation. Both fusions display oncogenic activities when introduced into Rat 2 cells in in vitro colony formation assays as compared to parental cells. The two fusions are also tumorigenic in vivo when implanted subcutaneously in NMRI nude mice, whereas parental cell do not grow in vivo. FGFR inhibition by the FGFR selective inhibitor Debio 1347 (CH5183284) reduced cell proliferation in vitro. In vivo sensitivity to FGFR inhibition could also be demonstrated for the two fusions. Altogether, these results suggest that these two novel oncogenic fusions are actionable targets in cholangiocarcinoma and that patients harboring these fusions could benefit from targeted FGFR inhibition, such as Debio 1347, currently investigated in a Phase I trial in selected patients harboring FGFR genetic alterations (NCT01948297). Citation Format: Anne Vaslin, Stefania Rigotti, Nathalie Lembrez, Gregoire Vuagniaux, Corinne Moulon, Hiroaki Tanaka. Characterization of two novel oncogenic FGFR2 fusions sensitive to the FGFR-selective inhibitor Debio 1347 in cholangiocarcinoma. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 689. doi:10.1158/1538-7445.AM2015-689
Cancer Research | 2014
Alexandra Oudot; Olivier Raguin; Lucile Bauché; Francis Bichat; Anne Vaslin; Hélène Maby-El Hajjami; Claudio Zanna; Grégoire Vuagniaux; Pierre Fumoleau; François Brunotte; Bertrand Collin
Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Introduction: Inhibitors of apoptosis proteins (IAPs) are key negative regulators of programmed cell death. Their frequent deregulation in most cancer types contributes to tumor cell survival and resistance to cancer therapy, making IAPs attractive therapeutic targets. Debio 1143 (formerly named AT-406), a new potent orally-available monovalent SMAC mimetic, targets multiple IAP members and is currently in clinical trials for cancer treatment. In this study, pharmaco-imaging was used to evaluate the effects of Debio 1143 on tumor cell death and metabolism in the triple negative breast cancer (TNBC) cell line MDA-MB-231. Material and Methods: The effects of Debio 1143 on caspase-3 activation (FACS), apoptosis-induced membrane changes (FACS and radioactive detection of Annexin V binding), and cell proliferation (MTS assay) were evaluated in MDA-MB-231 cells. In vivo pharmaco-imaging experiments were performed in CB17 SCID mice bearing subcutaneous MDA-MB-231 TNBC tumors. 99mTc-Annexin V SPECT/CT (Single Photon Emission Computed Tomography/Computed Tomography) imaging was performed 6 hours and 24 hours after a single treatment with vehicle, Debio 1143 (100 mg/kg per os) or paclitaxel (7.5 mg/kg intravenously), and was completed by gamma counting of organs. Tumor metabolism was evaluated by 18F-FDG (fluorodeoxyglucose) PET/CT (Positron Emission Tomography) while animals received repeated administrations of vehicle, Debio 1143 (100 mg/kg per os) or paclitaxel (7.5 mg/kg intravenously). Results: In MDA-MB-231 cells, Debio 1143 as a single agent was highly effective in inducing apoptosis. This was reflected by a dose-dependent activation of caspase-3, an increase of Annexin V cell binding, and a cytotoxic activity with a mean IC50 of 224 nM. In MDA-MB-231 tumor-bearing mice, Debio 1143 showed an effect on 99mTc-Annexin V tumor binding with an increased tumor SPECT signal and gamma counting results 6 hours after oral administration, while paclitaxel maximum effects were detected at 24 hours post-treatment. During repeated administration, oral Debio 1143 inhibited tumor growth, which was associated with a decreased tumor 18F-FDG uptake when measured during treatment (after 1 and 2 weeks of treatment). The effects on tumor growth and 18F-FDG uptake were still present 1 week after the end of the treatment period. Conclusion: This pharmaco-imaging study of the effects of Debio 1143 in TNBC showed that Debio 1143 induced apoptosis both in vitro and in vivo. Moreover, 18F-FDG PET imaging made it possible to observe the effect of Debio 1143, suggesting that this imaging technique may be useful in a clinical setting. To conclude, this study supports the use of Debio 1143 in the treatment of TNBC - a still unmet medical need. Citation Format: Alexandra Oudot, Olivier Raguin, Lucile Bauche, Francis Bichat, Anne Vaslin, Helene Maby-El Hajjami, Claudio Zanna, Gregoire Vuagniaux, Pierre Fumoleau, Francois Brunotte, Bertrand Collin. Pharmaco imaging study of the effects of Debio 1143, a new orally available IAP inhibitor, in a triple negative breast cancer model. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2757. doi:10.1158/1538-7445.AM2014-2757
Cancer Research | 2014
Marie Serova; Annemilai Tijeras-Raballand; Sebastien Albert; Sandrine Faivre; Eric Raymond; Anne Vaslin; Claudio Zanna; Grégoire Vuagniaux; Armand de Gramont
Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Background: Resistance to radiotherapy and chemotherapy-induced apoptosis is a hallmark of cancer. Inhibitors of apoptosis proteins (IAPs) are negative modulators of apoptosis frequently expressed in various cancers, and, as such, attractive targets to overcome resistance to cancer therapy. The oral SMAC mimetic Debio 1143 (a.k.a AT-406), an antagonist of multiple IAPs (cIAP1/2 and XIAP), is currently investigated in a Phase I oncology clinical trial. This study evaluated Debio 1143 activity as a single agent, and in combination with TNF-α, TRAIL, cisplatin or carboplatin, in various tumor models of squamous cell carcinoma of the head and neck (SCCHN). Materials and Methods: The antiproliferative effect of Debio 1143 was evaluated by MTT assay in human SCCHN cell lines SQ20B, SCC61, Hep2 and Detroit 562. The extent of apoptotic cell death was characterized by Western blot for cleaved caspase 3 and PARP. Tumor samples from SCCHN patients were surgically resected and cut into 300 µm thick slices using a tissue slicer (TIPCAN®). Each slice was exposed to 10 µM Debio 1143 and/or 1 µM of platinum-based drug for 48 hours. Tumor explants from 9 SCCHN patients were analyzed by immunohistochemistry or immunofluorescence to visualize the effects of treatment on various biomarkers of cell apoptosis, proliferation and drug target engagement. Results: Debio 1143 alone displayed limited antiproliferative activity in SCCHN cell lines. The addition of TRAIL or TNF-α potentiated the antiproliferative effects of Debio 1143 in 1 cell line (SQ20B) and in 3 cell lines (SQ20B, SCC61 and Detroit 562), respectively. Combining Debio 1143 with TNF-α induced cleavage of caspase 3 and PARP, suggesting induction of apoptosis. Exposure of fresh SCCHN tumor explants to Debio 1143 reduced c-IAP1 staining. Combination of Debio 1143 with cisplatin or carboplatin induced caspase 3 activation, suggesting apoptosis. Large necrotic areas were also found on tissue samples after combination therapy. Conclusion: In 3 out of 4 SCCHN cell lines, Debio 1143 potentiated TNF-α or TRAIL-induced antiproliferative effects. Debio 1143 combined with carboplatin and cisplatin induced caspase 3-dependent apoptosis in SCCHN patient tumor samples. Debio 1143 in combination with conventional chemotherapies could be a potential treatment for SCCHN patients. Citation Format: Marie Serova, Annemilai Tijeras-Raballand, Sebastien Albert, Sandrine Faivre, Eric Raymond, Anne Vaslin, Claudio Zanna, Gregoire Vuagniaux, Armand de Gramont. Effects of Debio 1143, a novel oral IAP inhibitor, in monotherapy and in combination with platinum drugs in human SCCHN tumor specimens. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2752. doi:10.1158/1538-7445.AM2014-2752
Molecular Cancer Therapeutics | 2013
Marie Serova; Annemilai Tijeras-Raballand; Sebastien Albert; Sandrine Faivre; Eric Raymond; Anne Vaslin; Claudio Zanna; Gregoire Vuagniaux; Armand de Gramont
Background: Resistance to radiotherapy and chemotherapy-induced apoptosis is a hallmark of cancer. Inhibitors of apoptosis proteins (IAPs) are negative modulators of apoptosis frequently expressed in various cancers, and, as such, attractive targets to overcome resistance to cancer therapy. The oral SMAC mimetic Debio 1143 (D1143, a.k.a AT-406), an antagonist of multiple IAPs (cIAP1/2 and XIAP), is currently investigated in a Phase I oncology clinical trial. This study aimed at evaluating D1143 activity as a single agent and in combination with TNF-α, TRAIL, cisplatin or carboplatin, in various SCCHN models. Materials and Methods: The antiproliferative effects of D1143 were evaluated in a panel of 5 human SCCHN cell lines by MTT assay. Baseline and phosphorylated protein levels were detected by Western blot analysis. Tumor samples from SCCHN patients were surgically resected and cut into 300 µm thick slices using a tissue slicer (TIPCAN®). Each slice was exposed to 10 µM D1143 and/or 1 µM of platinum-based drug for 48 hours. Tumor samples were analyzed by immunohistochemistry (IHC) or immunofluorescence to visualize the effects of the treatment on various biomarkers of cell apoptosis, proliferation, and drug target engagement. Results: A panel of 5 SCCHN cell lines was characterized for the expression of c-IAP1/2, XIAP, Bcl2, LRIG1, and other proteins implicated in resistance to cell death. D1143 alone displayed limited antiproliferative activity in only one cell line (Detroit 562). However, two SCCHN cell lines were sensitive to D1143 and TRAIL combined (SQ20B and SCC15), three were sensitive to D1143 and TNF-α combined (SQ20B, SCC61, and Detroit 562), and one was resistant to both combinations (HEP2). In Detroit 562 sensitive cells, 10 µM D1143 induced sustained cIAP1/2 degradation after 15 minutes of exposure. In contrast, in the HEP2 insensitive cell line, D1143 induced slight and transient inhibition of cIAP1/2. IHC analyses revealed that ex vivo exposure to D1143 of tumor explants freshly resected from SCCHN patients decreased cIAP1 staining. Treatment of tumor samples with D1143 combined with cisplatin or carboplatin augmented the cleavage of caspase 3 compared to controls suggesting induction of apoptosis. Conclusion: In 4 out of 5 SCCHN cell lines, D1143 induced cIAP1/2 degradation and potentiated TNF-α or TRAIL-induced antiproliferative effects. D1143 combined with carboplatin and cisplatin in SCCHN patient samples induced caspase 3-dependent apoptosis. D1143 in combination with conventional chemotherapies may be considered as a potential treatment for SCCHN patients. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):C20. Citation Format: Marie Serova, Annemilai Tijeras-Raballand, Sebastien Albert, Sandrine Faivre, Eric Raymond, Anne Vaslin, Claudio Zanna, Gregoire Vuagniaux, Armand de Gramont. In vitro effects of Debio 1143, a novel oral IAP inhibitor, in human SCCHN cell lines and tumor specimens. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr C20.
Cancer Research | 2013
D. Viertl; Florence Perillo-Adamer; Stefania Rigotti; Jean-Luc Muralti; Hélène Maby-El Hajjami; Anne Vaslin; Grégoire Vuagniaux; Oscar Matzinger
Purpose/objectives: Resistance of tumor cells to chemotherapy (CT) or radiotherapy (RT)-induced apoptosis is a major problem in the treatment of head and neck squamous cell carcinoma (HNSCC), and highlights the need for new therapeutic strategies. The small molecule Debio 1143 (D1143) (a.k.a. SM-406 or AT-406) is a potent orally-active, monovalent Smac-mimetic designed to promote programmed cell death in tumor cells by blocking the activity of inhibitor of apoptosis proteins (IAPs) to create conditions in which apoptosis can proceed. This study aimed to test the efficacy of D1143 as a single agent and/or in combination with CT or RT in HNSCC models. Materials and methods: The effect of D1143 was assessed by the colony forming assay on a panel of HNSCC patient-derived xenograft (PDX) tumors and established cell lines. Tumor cells were incubated at various concentrations of D1143 (ranging from 0.1 to 30 μM) and treated with increasing doses of radiation or cisplatin. Cells were cultured for a minimum of 10 days before being fixed and stained. The number of surviving colonies was then recorded and compared to an untreated group. In parallel, various treatment sequences were tested, (before, during or post-irradiation) to determine the effect of D1143 on primary apoptosis (early, pre-mitotic apoptosis) and secondary apoptosis (late, post-mitotic reproductive cell death). The in vivo chemo/radiosensitizing effect of oral treatment with D1143 was further evaluated in nude mice bearing HNSCC tumor xenografts. Results: D1143 alone selectively inhibited colony formation of several PDX tumors and established cell lines. In addition, combination experiments found D1143 to be highly effective in enhancing cell death induced by RT or cisplatin. A synergistic effect of D1143 was observed in the majority of the tested cell lines treated by RT, whereas D1143 enhanced the activity of cisplatin in a subset of tumor cells. Interestingly, the radiosensitizing effect of D1143 was preeminent when cells were co-incubated with D1143 24 hours after irradiation, showing that D1143 efficiently impacts late apoptosis due to mitotic catastrophe and/or other cell death events that arise after irradiation. In mice bearing HNSCC radio-resistant tumors, D1143 given orally in combination with RT delayed tumor growth for more than 40 days when compared to RT only and for more than 60 days when compared to vehicle only. In addition, oral administration of D1143 showed additive to synergistic activity when combined with intravenous cisplatin in nude mice bearing PDX HNSCC tumors. Conclusion: Altogether, these results show that D1143 exhibits activity as a single agent and can potentiate the effects of chemo/radiotherapy in HNSCC models indicating that D1143 has a promising therapeutic potential for the treatment of HNSCC. Citation Format: David Viertl, Florence Perillo-Adamer, Stefania Rigotti, Jean-Luc Muralti, Helene Maby-El Hajjami, Anne Vaslin, Gregoire Vuagniaux, Oscar Matzinger. The SMAC-mimetic Debio 1143 efficiently enhanced chemo and radiotherapy in head and neck squamous cell carcinoma models. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2055. doi:10.1158/1538-7445.AM2013-2055
Journal of Clinical Oncology | 2017
Claudio Zanna; Anne Vaslin; Martin H. Voss; Rebecca S. Heist; Cinta Hierro; Jeffrey G. Supko; Kiyohiko Nakai; Nobuya Ishii; Annick Menetrey; Marie-Claude Roubaudi-Fraschini; Julien Cordier; Jerome Douchain; Valerie Nicolas-Metral
Journal of Clinical Oncology | 2017
Martin H. Voss; Cinta Hierro; Rebecca S. Heist; James M. Cleary; Funda Meric-Bernstam; Leena Gandhi; Nobuya Ishii; Yulia Kirpicheva; Valerie Nicolas-Metral; Anna Pokorska-Bocci; Daniela Purcea; Anne Vaslin; Corinne Moulon; Claudio Zanna; Keith T. Flaherty; Josep Tabernero; José Baselga